Next Article in Journal
Formulation Development of a COVID-19 Recombinant Spike Protein-Based Vaccine
Previous Article in Journal
Gold Nanoparticle Virus-like Particles Presenting SARS-CoV-2 Spike Protein: Synthesis, Biophysical Properties and Immunogenicity in BALB/c Mice
Previous Article in Special Issue
The Effect of Previous Exposure to Malaria Infection and Clinical Malaria Episodes on the Immune Response to the Two-Dose Ad26.ZEBOV, MVA-BN-Filo Ebola Vaccine Regimen
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Immunogenicity of an Extended Dose Interval for the Ad26.ZEBOV, MVA-BN-Filo Ebola Vaccine Regimen in Adults and Children in the Democratic Republic of the Congo

by
Edward Man-Lik Choi
1,*,†,
Kambale Kasonia
1,†,
Hugo Kavunga-Membo
2,
Daniel Mukadi-Bamuleka
2,
Aboubacar Soumah
3,
Zephyrin Mossoko
2,
Tansy Edwards
4,5,
Darius Tetsa-Tata
1,
Rockyath Makarimi
3,
Oumar Toure
3,
Grace Mambula
3,
Hannah Brindle
1,
Anton Camacho
3,
Nicholas E. Connor
1,
Pierre Mukadi
2,
Chelsea McLean
6,
Babajide Keshinro
6,
Auguste Gaddah
7,
Cynthia Robinson
6,
Kerstin Luhn
6,
Julie Foster
1,
Chrissy h. Roberts
1,
John Emery Johnson
8,
Nathalie Imbault
9,
Daniel G. Bausch
1,10,
Rebecca F. Grais
3,
Deborah Watson-Jones
1,11 and
Jean Jacques Muyembe-Tamfum
2 on behalf of the Tujiokowe Study Consortium
add Show full author list remove Hide full author list
1
Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London WC1E 7HT, UK
2
Institut National de Recherche Biomédicale, Kinshasa P.O. Box 1192, Democratic Republic of the Congo
3
Epicentre, 75019 Paris, France
4
MRC International Statistics and Epidemiology Group, Faculty of Epidemiology and Population Health, London School of Hygiene & Tropical Medicine, London WC1E 7HT, UK
5
School of Tropical Medicine and Global Health, Nagasaki University, Nagasaki 852-8523, Japan
6
Janssen Vaccines and Prevention B.V., 2333 CN Leiden, The Netherlands
7
Janssen Research & Development, 2340 Beerse, Belgium
8
Médecins Sans Frontières, 75019 Paris, France
9
Coalition for Epidemic Preparedness Innovations, 0191 Oslo, Norway
10
FIND, 1218 Geneva, Switzerland
11
Mwanza Intervention Trials Unit, National Institute for Medical Research, Mwanza P.O. Box 11936, Tanzania
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Vaccines 2024, 12(8), 828; https://doi.org/10.3390/vaccines12080828
Submission received: 25 May 2024 / Revised: 21 June 2024 / Accepted: 18 July 2024 / Published: 23 July 2024

Abstract

:
During the 2018–2020 Ebola virus disease outbreak in Democratic Republic of the Congo, a phase 3 trial of the Ad26.ZEBOV, MVA-BN-Filo Ebola vaccine (DRC-EB-001) commenced in Goma, with participants being offered the two-dose regimen given 56 days apart. Suspension of trial activities in 2020 due to the COVID-19 pandemic led to some participants receiving a late dose 2 outside the planned interval. Blood samples were collected from adults, adolescents, and children prior to their delayed dose 2 vaccination and 21 days after, and tested for IgG binding antibodies against Ebola virus glycoprotein using the Filovirus Animal Nonclinical Group (FANG) ELISA. Results from 133 participants showed a median two-dose interval of 9.3 months. The pre-dose 2 antibody geometric mean concentration (GMC) was 217 ELISA Units (EU)/mL (95% CI 157; 301) in adults, 378 EU/mL (281; 510) in adolescents, and 558 EU/mL (471; 661) in children. At 21 days post-dose 2, the GMC increased to 22,194 EU/mL (16,726; 29,449) in adults, 37,896 EU/mL (29,985; 47,893) in adolescents, and 34,652 EU/mL (27,906; 43,028) in children. Participants receiving a delayed dose 2 had a higher GMC at 21 days post-dose 2 than those who received a standard 56-day regimen in other African trials, but similar to those who received the regimen with an extended interval.

1. Introduction

From 2018 to 2020, the Democratic Republic of the Congo (DRC) experienced its tenth Ebola virus disease (EVD) epidemic, with the epicentre in North Kivu Province [1]. Despite control efforts, including ring-vaccination of EVD case contacts and contacts of contacts using the rVSV-ZEBOV-GP vaccine (Ervebo®, Merck & Co., Inc., Whitehouse Station, NJ, USA) [2], new cases continued to emerge, and this outbreak became the largest EVD outbreak in the DRC, and second largest on record. In May 2019, the WHO Strategic Advisory Group of Experts on Immunization (SAGE) recommended deploying a second investigational EVD vaccine to populations at some risk of exposure with provision of informed consent [3].
In November 2019, the DRC Ministry of Health, together with consortium partners, implemented a large-scale, open-label, non-randomised phase 3 trial (DRC-EB-001) that planned to vaccinate up to 500,000 healthy eligible individuals aged one year or above in North Kivu. The study began in the city of Goma and aimed to assess the vaccine effectiveness of the adenovirus serotype 26 vector-based vaccine encoding the Ebola virus (EBOV) Mayinga glycoprotein (Ad26.ZEBOV) as dose 1 and the modified Vaccinia Ankara Bavarian Nordic vector-based vaccine encoding multiple filovirus proteins (MVA-BN-Filo) as dose 2, with the two doses given at a recommended interval of 56 days (window −14/+28 days) [4,5].
In a stringent animal model, the Ad26.ZEBOV, MVA-BN-Filo regimen with a 56-day interval protected vaccinated non-human primates from lethal EBOV challenges [6]. In previous phase 1 and phase 2 trials, this regimen with a 56-day interval produced stronger binding and neutralising antibody responses than regimens using shorter intervals (14 or 28 days) [7,8,9,10]. Vaccine immunogenicity did not diminish when adult volunteers received dose 2 more than 56 days after dose 1 [8,9,11]. However, there are no data on delayed dose 2 vaccination amongst children.
The DRC-EB-001 trial enrolled and vaccinated over 20,000 participants with dose 1 within the first four months. As COVID-19 became a pandemic, the trial suspended vaccination activities in April 2020, which were not resumed until five months later. In addition, the North Kivu EVD epidemic was declared over in June 2020 [12]. While the lack of confirmed EVD cases in the study area made it impossible to evaluate vaccine effectiveness, the delayed dose 2 vaccination of over 6000 participants dictated by the COVID-19 pandemic presented an opportunity to explore immune responses to the Ad26.ZEBOV, MVA-BN-Filo regimen outside the recommended 56-day interval. After discussion and approval from the trial’s Data Safety Monitoring Board, the consortium agreed to investigate the impact of the dose 2 delay on vaccine immunogenicity in adults, adolescents, and children, and to compare the results with earlier trials that used the same two-dose regimen. The results are presented here.

2. Materials and Methods

2.1. Study Design

This study is an exploratory component of the DRC-EB-001 phase 3 trial. The main trial is registered at Clinicaltrials.gov (NCT04152486). Full study methods for the main DRC-EB-001 trial have been previously described [4]. Key safety results on the full vaccinated cohort are presented in a companion paper to this manuscript [13].

2.2. Study Participants

From 30 October 2020 to 16 November 2020, DRC-EB-001 participants who returned to the six study vaccination centres in the morning for a delayed dose 2 vaccination were invited to join the immunogenicity subset (Figure 1). All immunogenicity visits were completed by 7 December 2020. The target population comprised at least 50 adult participants aged 18 years or over, approximately 25 adolescents aged 12–17 years and approximately 25 children aged 4–11 years. When study activities resumed in late 2020, participants were re-consented and assessed for eligibility to join this immunogenicity substudy prior to dose 2 vaccination. Individuals were deemed eligible if they met the original inclusion and exclusion criteria in the main study protocol, did not have any contraindication to dose 2, were reachable by phone, and were willing to donate venous blood samples at two visits (Supplementary Table S1) [4]. Individuals who had received another EVD vaccine (rVSV-ZEBOV-GP) and those who missed a Filovirus Animal Nonclinical Group (FANG) ELISA result for one or more time points were excluded from immunogenicity analysis. The DRC-EB-001 trial did not require any safety laboratory assessments as a screening step for inclusion.

2.3. Procedures

Venous blood samples for the determination of humoral immune responses were collected prior to vaccination on the day of the dose 2 visit and 21 days post-dose 2 (window −3/+7 days). The sample volume was 5 mL per visit for participants aged six years or over, and 2.5 mL per visit for participants aged four and five years. The blood samples were processed at the Institut National de la Recherche Biomédicale laboratory in Goma. Frozen serum aliquots were sent to the Q2 Solutions Laboratory (San Juan Capistrano, CA, USA) for immunogenicity evaluations [14].

2.4. Vaccines

Ad26.ZEBOV (Zabdeno®, Janssen Pharmaceutica, Beerse, Belgium) is an adenovirus serotype 26 (Ad26)-vectored vaccine, encoding the EBOV Mayinga glycoprotein (GP) and given as an intramuscular injection of 0.5 mL liquid suspension, containing 5 × 1010 virus particles [15]. MVA-BN-Filo (Mvabea®, Janssen Pharmaceutica, Beerse, Belgium) is a recombinant modified Vaccinia Ankara (MVA)-vectored vaccine which is highly attenuated and does not replicate in human cells. It is multivalent, encoding the GP of EBOV (100% homologous to the EBOV GP in Ad26.ZEBOV), Sudan, and Marburg Musoke viruses, and the nucleoprotein of Taï Forest virus [16]. MVA-BN-Filo was given as an intramuscular injection of 0.5 mL liquid suspension, containing 1 × 108 infectious units. Ad26.ZEBOV was manufactured by IDT Biologika GmbH, Dessau-Roßlau, Germany, and MVA-BN-Filo by Bavarian Nordic A/S, Kvistgård, Denmark, for Janssen Vaccines & Prevention B.V., Leiden, The Netherlands. Neither vaccine contains any adjuvant [4,13].

2.5. Immunogenicity Evaluations

Antibody responses against EBOV GP were measured by the FANG enzyme-linked immunosorbent assay (ELISA) at Q2 Solutions Laboratory (San Juan Capistrano, CA, USA). This assay has been validated for testing human serum [14]. The concentrations of IgG antibodies binding to recombinant EBOV GP were presented as geometric mean concentrations (GMCs) in ELISA Units (EU)/mL. The lower limit of quantification (LLOQ) for this ELISA, as determined in the assay validation, was 36.11 EU/mL. Samples with results above the upper limit of quantification (ULOQ) were diluted in assay diluent and retested. The retesting results were multiplied by the pre-dilution factor to determine a final result.

2.6. Data Management

Participant data were collected on a combination of paper and electronic case report forms (CRFs). Data collected on paper CRFs were double-entered into a REDCap database (version 11.0.3). Electronic CRFs completed on tablets using ODK (ODK Collect v1.16 to v1.22, ODK Aggregate v1.44 to v2.03) were encrypted before syncing to a secure server and imported into REDCap, as previously described [4]. Query resolution and quality assurance were performed on an ongoing basis by members of the data team, quality team and the internal monitoring team.

2.7. Statistical Analysis

The sample size calculations were based on estimating the GMC with precision with no formal statistical hypothesis testing considerations. We assumed a standard deviation of 0.55 (on log10-scale) of the binding antibody concentrations as measured by ELISA at 21 days post dose 2, based on the maximum observed standard deviation in the EBL3001 study plus 10% [11,17]. It was estimated that a sample size of 50 would allow estimation of a two-sided 95% confidence interval with a distance from the mean to the limits that is equal to 0.156 (on log10-scale).
Individuals with FANG ELISA results for both pre-dose 2 and 21 days post-dose 2 samples (i.e., paired samples) were included in the immunogenicity analysis population.
The age and sex distributions were described, along with the mean, standard deviation (SD), median, and minimum–maximum range of the time intervals between dose 1 and dose 2 in days and months (28 days). The GMC of antibodies and corresponding 95% confidence intervals (CI) were calculated per time point for all participants with paired samples within the defined age groups for children, adolescents, and adults.
Fold change (fold increase) of antibody concentrations was calculated as the ratio of the 21 days post-dose 2 to the pre-dose 2 GMC value on the original measurement scale before calculation of geometric mean fold change and corresponding 95% CIs for each age group. Differences in values at 21 days post-dose 2 by age group, conditional on pre-dose 2 (day 0) values, were tested using ANOVA, analysing data on the log base10 scale with the outcome as the 21 days post-dose 2 values, adjusting for pre-dose 2 (day 0) values. Graphical presentation of data included jittered boxplots to show the distribution of immunogenicity data by time and age group, and confidence interval plots to show summary data by time and age group. The LLOQ is shown as 36.11 and the ULOQ as 194,938.9. Results above the ULOQ were imputed as 194,939 EU/mL.
Because our DRC trial and other African trials of the same vaccine were conducted in different populations using different inclusion/exclusion criteria, no formal statistical testing of their comparison was performed.

2.8. Ethics Statement

The protocol was approved by the Ethics Committee/Institutional Review Board of the London School of Hygiene & Tropical Medicine (LSHTM EC; Ref 17471), the Médecins Sans Frontières Ethics (MSF-ERB; ID:1922), the University of Kinshasa School of Public Health (CE-ESP; ESP/CE/234/2019), and Comité National d’Ethique de la Santé (CNES; n°138/CNES/BN/PMMF2019) [4].

3. Results

3.1. Study Participants

In total, 161 individuals were screened and 150 gave consent and/or assent to participate in the subset and provided a blood sample prior to receiving their dose 2 vaccine (Figure 2). Seventeen participants were excluded from analysis, including two adults who had received rVSV-ZEBOV-GP in addition to the study vaccines (Supplementary Table S2), leaving a total of 133 individuals with paired pre- and post-dose 2 samples. The final Immunogenicity Analysis Population comprised 49 adults, 32 adolescents, and 52 children (Figure 2). All immunogenicity subset participants received their dose 2 outside the recommended interval of 56 days (window −14/+28 days) post-dose 1. The median (minimum–maximum) interval between dose 1 and dose 2 was 9.3 (8.7–12.1) months for adults, 9.5 (8.9–11.7) months for adolescents, and 9.1 (8.9–10.1) months for children. The baseline demographics of the participants included in the immunogenicity substudy are shown in Table 1.

3.2. Vaccine Immunogenicity

Upon administration of a delayed dose 2, the antibody GMC increased by 102-fold in adults, 100-fold in adolescents and 62-fold in children within 21 days (Figure 3, Table 2). In the absence of pre-dose 1 data, pre-dose 2 GMC in all participants served as a conservative baseline antibody level. All but one participant 132/133 (99%) had an antibody GMC that increased more than 2.5 times after receiving dose 2 and may thus be considered responders.
Amongst 133 paired samples, linear regression was used to model 21 days post-dose 2 data on the log10 scale, conditional on pre-dose 2 (day 0) values on the log base10 scale, to investigate differences in fold change (fold increase) by age group. The analysis showed post-dose 2 antibody values to be higher in adolescents compared to adults (regression coefficient: 0.175, 95% CI: 0.126, 0.338, p = 0.035), when accounting for pre-dose 2 values. However, there was no significant difference between children and adults (regression coefficient: 0.097, 95% CI: −0.056, 0.250, p = 0.214). More variability in pre-dose 2 values can be seen in the adult group (Supplementary Figure S1).
The antibody GMC following the Ad26.ZEBOV, MVA-BN-Filo regimen with an extended two-dose interval observed in our study was higher than that reported in other sub-Saharan African trials [8,11,17,18,19,20]. Immunogenicity data stratified by the same age groups are available from the EBL3001 trial (NCT02509494) conducted in Sierra Leone and the EBL2002 trial (NCT02564523) conducted in Burkina Faso, Côte D’Ivoire, Kenya, and Uganda. Both EBL3001 and EBL2002 measured anti-EBOV GP IgG binding antibody GMC at 21 days after dose 2 using the same FANG ELISA at the same FDA-approved central laboratory as this study, thus permitting a direct comparison between three trials. As shown in Table 3, the post-dose 2 GMC observed in this study was at least 3.4 times higher than among EBL3001 participants who received the standard 56-day regimen in all three age groups. Similarly, the GMC observed in this study was at least 2.0 times higher than in healthy EBL2002 participants who received the standard regimen. There is no overlap between the 95% CIs around GMCs in these comparisons.
The heightened immunogenicity induced by late dose 2 administration in this study is reminiscent of the out-of-window antibody results obtained in EBL2002. In that trial, 169 healthy adult participants also received dose 2 beyond the protocol-defined window due to a temporary study pause [8]. As the two-dose interval extended from 56 to ≥280 days among these EBL2002 adult participants, the 21 days post-dose 2 antibody GMC increased from 7518 (95% CI 6468; 8740 EU/mL) to 23,897 EU/mL (95% CI 16,703; 34,188) (Figure 4). The GMC reported among the ≥280 days out-of-window EBL2002 adults (median interval 295 days), at 23,897 EU/mL (95% CI 16,703; 34,188), is comparable to the GMC observed among adults in this substudy (median interval 259 days), at 22,194 EU/mL (95% CI 16,726; 29,449). In EBL2002, extending the two-dose interval from 56 to ≥280 days also increased the pre- and post-dose 2 GMC fold change from 21-fold to 113-fold. The positive correlation between the length of the interval and the post-vaccination GMC can be illustrated by plotting the EBL2002 healthy adult data and fitting a curve using ordinary least square linear regression (Figure 4). When the adult data from our substudy are added to the plot, the immunogenicity of the delayed dose 2 in DRC-EB-001 appear consistent with the out-of-window immunogenicity in EBL2002.

4. Discussion

We measured the immunogenicity of the Ad26.ZEBOV, MVA-BN-Filo vaccine regimen in participants who received a delayed dose 2 and showed that extending the two-dose interval from the standard 56 days to approximately nine months enhanced antibody responses in adults, adolescents, or children. The EBOV GP binding antibody concentrations observed in our study are significantly higher than those reported in the EBL2002 and EBL3001 trials in other sub-Saharan African countries using the standard 56-day regimen. Furthermore, the immunogenicity in this study is consistent with the out-of-window immunogenicity in EBL2002, suggesting that the high antibody concentrations observed in our study are the result of delaying dose 2 administration. Our result is similar to previous findings that longer intervals between first and second dose can increase immunogenicity of vaccines against influenza A, malaria, and COVID-19 [21,22,23]. In the case of ChAdOx1.S nCoV-19, the Oxford-AstraZeneca adenovirus-vectored COVID-19 vaccine, extending the two-dose interval from less than six weeks to over 12 weeks increased binding and neutralising antibody responses as well as vaccine efficacy in protection against symptomatic infection [24,25].
This study has several limitations. Pre-dose 1 vaccination blood samples were not collected because the immunogenicity subset was implemented as an exploratory objective after all dose 1 vaccinations had concluded. It was therefore not possible to measure the vaccine responder rate using pre-dose 1 baseline GMC values, as in previous studies. The immunogenicity subset of the DRC-EB-001 trial did not include a comparator arm that received the standard 56-day regimen. Since all participants in the immunogenicity subset received their dose 2 vaccination between 8 to 12 months post-dose 1, direct comparison of the immunogenicity of the standard dosing schedule and the extended interval with a delayed dose 2 could not be measured within the same trial population. Because the immunogenicity follow-up period was only 21 days (window −3/+7 days), long-term persistence of the EBOV antibodies following a delayed dose could not be measured. To date there are still no clinical data on the vaccine efficacy of the Ad26.ZEBOV, MVA-BN-Filo regimen. Nor have the mechanistic correlates of protection been established. Nevertheless, the regimen’s clinical benefits were inferred by immunobridging from a stringent animal model, where vaccinated non-human primates (NHP) were 100% protected against EBOV challenge [6]. In these experiments, NHP survival was strongly associated with anti-EBOV GP binding antibody GMC. Of note, the GMCs achieved 21 days after Ad26.ZEBOV, MVA-BN-Filo vaccination in studies that implemented the standard 56-day regimen and this study that investigated a delayed dose 2 were at least as high as those achieved 28 days after vaccination with rVSV-ZEBOV-GP (Ervebo®), which demonstrated 100% protection against EVD in a phase 3 trial [18,26,27].
Taken together, results from the DRC-EB-001 trial add to the existing literature to support deployment of the Ad26.ZEBOV, MVA-BN-Filo vaccine for EVD as prophylactic vaccines for at-risk populations. During EVD outbreaks, every effort should be made to ensure that the complete regimen is delivered within the recommended 56-day interval in order to reach potentially protective levels of antibodies as soon as possible. However, a host of logistical challenges are often encountered in these difficult settings which may affect routine access to vaccination, including insecurity, lack of trained healthcare workers, population movement, and difficult maintenance of the cold chain [28]. Indeed, many of these were encountered during the DRC-EB-001 trial. The knowledge that, when necessary, delayed administration of dose 2 of the Ad26.ZEBOV, MVA-BN-Filo regimen has no negative impact on immunogenicity, gives comfort to the use of the vaccine even in the most turbulent settings. Public health and immunisation services should be encouraged to complete the two-dose schedule even if this is with an extended second dose window.

5. Conclusions

In conclusion, the Ebola vaccine regimen Ad26.ZEBOV, MVA-BN-Filo induced strong antibody responses in DRC-EB-001 trial participants who received dose 2 outside the recommended interval of 56 days (window −14/+28 days) after dose 1. Based on comparison with previous studies, delaying dose 2 administration may increase the level of anti-EBOV GP binding IgG antibody in children, adolescents, and adults. Nevertheless, because a person is still vulnerable to EBOV infection after the first dose of vaccine, maintaining the 56-day interval is still recommended when possible.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/vaccines12080828/s1, Supplementary Table S1: Participant eligibility criteria; Supplementary Table S2: Participants who had previously received the rVSV-ZEBOV-GP Ebola vaccine; Supplementary Figure S1: Individual trajectories of Ebola virus glycoprotein-specific IgG binding antibody responses over time.

Author Contributions

E.M.-L.C. drafted the manuscript and conducted the literature search for references. D.W.-J., E.M.-L.C., T.E., K.K., H.K.-M., D.M.-B., C.h.R., C.R., B.K., A.G., C.M., K.L., A.S., Z.M., D.T.-T., O.T., G.M., R.M., H.B., A.C., N.E.C., P.M., J.F., J.E.J., N.I., D.G.B., R.F.G. and J.J.M.-T. were involved in the study concept and design, study conduct and interpretation of results. J.J.M.-T. was the clinical trial principal investigator (PI) at Institut National de la Recherche Biomédicale in the DRC. D.W.-J. and D.G.B. were the co-PIs at the London School of Hygiene & Tropical Medicine. R.F.G. was the lead investigator at Epicentre. J.E.J. was the project lead for MSF. C.R. was the lead scientist for the programme at Janssen Vaccines & Prevention B.V. E.M.-L.C. was the trial coordinator and K.K. was the trial manager at LSHTM. H.K.-M., A.S., O.T., Z.M., G.M., D.M.-B., K.K., R.F.G. and D.W.-J. contributed to enrolment and clinical care of participants and data collection. D.M.-B. and Z.M. were responsible for laboratory analysis and samples management. C.h.R., D.T.-T., A.C. and H.B. were responsible for data management. N.E.C. and P.M. were responsible for quality assurance. A.S. and R.M. were the internal monitors at Epicentre. T.E. conducted the statistical analysis. E.M.-L.C. and T.E. have accessed and verified the data. All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported through funds from the Coalition for Epidemic Preparedness Innovations (CEPI) [FELS1903] and the Paul G. Allen Family Foundation [12885]. This work received additional support from the UK Foreign, Commonwealth & Development Office (FCDO), Wellcome [220506/Z/20/Z] and the European Union’s Horizon 2020 research and innovation programme under grant agreement No 857935. This publication reflects only the authors’ view, and the European Commission is not responsible for any use that may be made of the information it contains. CEPI, Wellcome and FCDO advised on the study design and protocol. C.h.R. also received funding from the Department of Health and Social Care using UK Aid Funding as part of the UK Vaccine Network, and is managed by the National Institute for Health and Care Research [PR-OD-1017-20001]. The views expressed in this publication are those of the author(s) and not necessarily those of the Department of Health and Social Care. The vaccines for this study were donated by Janssen Vaccines and Prevention B.V.

Institutional Review Board Statement

The study was conducted in accordance with the Declaration of Helsinki, and approved by the following Ethics Committees (Institutional Review Boards) for studies involving humans: LSHTM Ethics committee (LSHTM EC; Ref 17471 on 15 October 2019), the Médecins Sans Frontières Ethics review board (MSF-ERB; ID:1922 on 15 October 2019), the University of Kinshasa School of Public Health Ethics Committee (CE-ESP; ESP/CE/234/2019 on 31 August 2019), and the Comité National d’Ethique de la Santé (CNES; n°138/CNES/BN/PMMF/2019 on 30 September 2019).

Informed Consent Statement

Informed consent was obtained from all participants involved in this study.

Data Availability Statement

Following publication of the primary and exploratory outcomes as detailed in the protocol, deidentified participant data will be made available upon request via LSHTM Data Compass, the London School of Hygiene & Tropical Medicine research data repository at http://datacompass.lshtm.ac.uk (accessed on 16 August 2023). Requests with a defined analysis plan can be sent via LSHTM Data Compass. We believe that the study methods and results are clearly documented in this article. The clinical study protocol has been published with open access [4].

Acknowledgments

We thank the study teams of the vaccination centres; the management teams of the project office; the clinical trial monitors and the LSHTM management and administration teams. We thank Robert Kanwagi and the World Vision team for their efforts in community engagement. We thank all DRC-EB-001 study participants and their families.

Conflicts of Interest

The funders advised on the study design and protocol, but had no role in the collection, analyses, or interpretation of data; in the writing of the manuscript; or in the decision to publish the results. Janssen Vaccines & Prevention B.V. was the vaccine manufacturer and donated the vaccine for this study. B.K., A.G., C.M., K.L., and C.R. were full-time employees of Janssen, Pharmaceutical Companies of Johnson & Johnson at the time of the study. A.G., C.M., K.L., and C.R. declared ownership of shares in Janssen, Pharmaceutical Companies of Johnson & Johnson. D.W.-J. and Janssen Vaccines & Prevention B.V. are consortium members for the EBOVAC1 and EBOVAC3 projects. All other authors declare no conflicts of interests.

References

  1. World Health Organization. Ebola Outbreak 2018–2020 North Kivu-Ituri. 2020. Available online: https://www.who.int/emergencies/situations/Ebola-2019-drc- (accessed on 16 August 2023).
  2. European Medicines Agency. Ervebo Ebola Zaire Vaccine (rVSVG-ZEBOV-GP, Live). 12 December 2019. Available online: https://www.ema.europa.eu/en/medicines/human/EPAR/ervebo (accessed on 16 August 2023).
  3. World Health Organization. Strategic Advisory Group of Experts (SAGE) on Immunization Interim Recommendations on Vaccination against Ebola Virus Disease (EVD); World Health Organization: Geneva, Switzerland, 7 May 2019; Available online: https://cdn.who.int/media/docs/default-source/immunization/ebola/interim-ebola-recommendations-may-2019.pdf (accessed on 16 August 2023).
  4. Watson-Jones, D.; Kavunga-Membo, H.; Grais, R.F.; Ahuka, S.; Roberts, N.; Edmunds, W.J.; Choi, E.M.; Roberts, C.H.; Edwards, T.; Camacho, A.; et al. Protocol for a phase 3 trial to evaluate the effectiveness and safety of a heterologous, two-dose vaccine for Ebola virus disease in the Democratic Republic of the Congo. BMJ Open 2022, 12, e055596. [Google Scholar] [CrossRef]
  5. Pearson, C.A.B.; Edmunds, W.J.; Hladish, T.J.; Eggo, R.M. Potential test-negative design study bias in outbreak settings: Application to Ebola vaccination in Democratic Republic of Congo. Int. J. Epidemiol. 2022, 51, 265–278. [Google Scholar] [CrossRef]
  6. Roozendaal, R.; Hendriks, J.; van Effelterre, T.; Spiessens, B.; Dekking, L.; Solforosi, L.; Czapska-Casey, D.; Bockstal, V.; Stoop, J.; Splinter, D.; et al. Nonhuman primate to human immunobridging to infer the protective effect of an Ebola virus vaccine candidate. NPJ Vaccines 2020, 5, 112. [Google Scholar] [CrossRef]
  7. Goldstein, N.; Bockstal, V.; Bart, S.; Luhn, K.; Robinson, C.; Gaddah, A.; Callendret, B.; Douoguih, M. Safety and Immunogenicity of Heterologous and Homologous 2-Dose Regimens of Adenovirus Serotype 26– and Modified Vaccinia Ankara–Vectored Ebola Vaccines: A Randomized, Controlled Phase 1 Study. J. Infect. Dis. 2020, 226, 595–607. [Google Scholar] [CrossRef]
  8. Barry, H.; Mutua, G.; Kibuuka, H.; Anywaine, Z.; Sirima, S.B.; Meda, N.; Anzala, O.; Eholie, S.; Bétard, C.; Richert, L.; et al. Safety and immunogenicity of 2-dose heterologous Ad26.ZEBOV, MVA-BN-Filo Ebola vaccination in healthy and HIV-infected adults: A randomised, placebo-controlled Phase II clinical trial in Africa. PLoS Med. 2021, 18, e1003813. [Google Scholar] [CrossRef]
  9. Pollard, A.J.; Launay, O.; Lelievre, J.D.; Lacabaratz, C.; Grande, S.; Goldstein, N.; Robinson, C.; Gaddah, A.; Bockstal, V.; Wiedemann, A.; et al. Safety and immunogenicity of a two-dose heterologous Ad26.ZEBOV and MVA-BN-Filo Ebola vaccine regimen in adults in Europe (EBOVAC2): A randomised, observer-blind, participant-blind, placebo-controlled, phase 2 trial. Lancet Infect. Dis. 2021, 21, 493–506. [Google Scholar] [CrossRef]
  10. Milligan, I.D.; Gibani, M.M.; Sewell, R.; Clutterbuck, E.A.; Campbell, D.; Plested, E.; Nuthall, E.; Voysey, M.; Silva-Reyes, L.; McElrath, M.J.; et al. Safety and Immunogenicity of Novel Adenovirus Type 26- and Modified Vaccinia Ankara-Vectored Ebola Vaccines: A Randomized Clinical Trial. JAMA 2016, 315, 1610–1623. [Google Scholar] [CrossRef]
  11. Ishola, D.; Manno, D.; Afolabi, M.O.; Keshinro, B.; Bockstal, V.; Rogers, B.; Owusu-Kyei, K.; Serry-Bangura, A.; Swaray, I.; Lowe, B.; et al. Safety and long-term immunogenicity of the two-dose heterologous Ad26.ZEBOV and MVA-BN-Filo Ebola vaccine regimen in adults in Sierra Leone: A combined open-label, non-randomised stage 1, and a randomised, double-blind, controlled stage 2 trial. Lancet Infect. Dis. 2022, 22, 97–109. [Google Scholar] [CrossRef]
  12. World Health Organization. 10th Ebola Outbreak in the Democratic Republic of the Congo Declared Over; Vigilance against Flare-Ups and Support for Survivors Must Continue. 25 June 2020. Available online: https://www.who.int/news/item/25-06-2020-10th-ebola-outbreak-in-the-democratic-republic-of-the-congo-declared-over-vigilance-against-flare-ups-and-support-for-survivors-must-continue (accessed on 27 June 2024).
  13. Kavunga-Membo, H.; Watson-Jones, D.; Kasonia, K.; Edwards, T.; Camacho, A.; Mambula, G.; Tetsa-Tata, D.; Choi, E.M.-L.; Aboubacar, S.; Brindle, H.; et al. Delivery and Safety of a Two-Dose Preventive Ebola Virus Disease Vaccine in Pregnant and Non-Pregnant Participants during an Outbreak in the Democratic Republic of the Congo. Vaccines 2024, 12, 825. [Google Scholar] [CrossRef]
  14. Rudge, T.L., Jr.; Sankovich, K.A.; Niemuth, N.A.; Anderson, M.S.; Badorrek, C.S.; Skomrock, N.D.; Cirimotich, C.M.; Sabourin, C.L. Development, qualification, and validation of the Filovirus Animal Nonclinical Group anti-Ebola virus glycoprotein immunoglobulin G enzyme-linked immunosorbent assay for human serum samples. PLoS ONE 2019, 14, e0215457. [Google Scholar] [CrossRef]
  15. European Medicines Agency. Zabdeno Ebola Vaccine (Ad26.ZEBOV-GP [Recombinant]). 23 July 2020. Available online: https://www.ema.europa.eu/en/medicines/human/EPAR/zabdeno (accessed on 16 August 2023).
  16. European Medicines Agency. Mvabea Ebola Vaccine (MVA-BN-Filo [Recombinant]). 23 July 2020. Available online: https://www.ema.europa.eu/en/medicines/human/EPAR/mvabea (accessed on 16 August 2023).
  17. Afolabi, M.I.D.; Manno, D.; Keshinro, B.; Bockstal, V.; Rogers, B.; Owusu-Kyei, K.; Serry-Bangura, A.; Swaray, I.; Lowe, B.; on behalf of the EBL3001 Study Group; et al. Safety and immunogenicity of a two-dose Ad26. ZEBOV, MVA-BN-Filo Ebola vaccine in children: A randomised, double-blind, controlled trial in Sierra Leone. Lancet Infect. Dis. 2021, 22, 110–122. [Google Scholar] [CrossRef]
  18. PREVAC Study Team. Randomized Trial of Vaccines for Zaire Ebola Virus disease. N. Engl. J. Med. 2022, 387, 2411–2424. [Google Scholar] [CrossRef] [PubMed]
  19. Anywaine, Z.; Barry, H.; Anzala, O.; Mutua, G.; Sirima, S.B.; Eholie, S.; Kibuuka, H.; Bétard, C.; Richert, L.; Lacabaratz, C.; et al. Safety and immunogenicity of 2-dose heterologous Ad26.ZEBOV, MVA-BN-Filo Ebola vaccination in children and adolescents in Africa: A randomised, placebo-controlled, multicentre Phase II clinical trial. PLoS Med. 2022, 19, e1003865. [Google Scholar] [CrossRef]
  20. Larivière, Y.; Matuvanga, T.Z.; Osang’ir, B.I.; Milolo, S.; Meta, R.; Kimbulu, P.; Robinson, C.; Katwere, M.; McLean, C.; Lemey, G.; et al. Ad26.ZEBOV, MVA-BN-Filo Ebola virus disease vaccine regimen plus Ad26.ZEBOV booster at 1 year versus 2 years in health-care and front-line workers in the Democratic Republic of the Congo: Secondary and exploratory outcomes of an open-label, randomised, phase 2 trial. Lancet Infect. Dis. 2024, 24, 746–759. [Google Scholar]
  21. Ledgerwood, J.E.; Zephir, K.; Hu, Z.; Wei, C.-J.; Chang, L.; Enama, M.E.; Hendel, C.S.; Sitar, S.; Bailer, R.T.; Koup, R.A.; et al. Prime-boost interval matters: A randomized phase 1 study to identify the minimum interval necessary to observe the H5 DNA influenza vaccine priming effect. J. Infect. Dis. 2013, 208, 418–422. [Google Scholar] [CrossRef] [PubMed]
  22. Fernandez-Arias, C.; Arias, C.F.; Zhang, M.; Herrero, M.A.; Acosta, F.J.; Tsuji, M. Modeling the effect of boost timing in murine irradiated sporozoite prime-boost vaccines. PLoS ONE 2018, 13, e0190940. [Google Scholar] [CrossRef]
  23. Flaxman, A.; Marchevsky, N.G.; Jenkins, D.; Aboagye, J.; Aley, P.K.; Angus, B.; Belij-Rammerstorfer, S.; Bibi, S.; Bittaye, M.; Cappuccini, F.; et al. Reactogenicity and immunogenicity after a late second dose or a third dose of ChAdOx1 nCoV-19 in the UK: A substudy of two randomised controlled trials (COV001 and COV002). Lancet 2021, 398, 981–990. [Google Scholar] [CrossRef]
  24. Voysey, M.; Clemens, S.A.C.; Madhi, S.A.; Weckx, L.Y.; Folegatti, P.M.; Aley, P.K.; Angus, B.; Baillie, V.L.; Barnabas, S.L.; Bhorat, Q.E.; et al. Single-dose administration and the influence of the timing of the booster dose on immunogenicity and efficacy of ChAdOx1 nCoV-19 (AZD1222) vaccine: A pooled analysis of four randomised trials. Lancet 2021, 397, 881–891. [Google Scholar] [CrossRef] [PubMed]
  25. Rogawski McQuade, E.T.; Breskin, A. Longer intervals and extra doses of ChAdOx1 nCoV-19 vaccine. Lancet 2021, 398, 933–935. [Google Scholar] [CrossRef]
  26. Henao-Restrepo, A.M.; Longini, I.M.; Egger, M.; Dean, N.E.; Edmunds, W.J.; Camacho, A.; Carroll, M.W.; Doumbia, M.; Draguez, B.; Duraffour, S.; et al. Efficacy and effectiveness of an rVSV-vectored vaccine expressing Ebola surface glycoprotein: Interim results from the Guinea ring vaccination cluster-randomised trial. Lancet 2015, 386, 857–866. [Google Scholar] [CrossRef]
  27. Henao-Restrepo, A.M.; Camacho, A.; Longini, I.M.; Watson, C.H.; Edmunds, W.J.; Egger, M.; Carroll, M.W.; Dean, N.E.; Diatta, I.; Doumbia, M.; et al. Efficacy and effectiveness of an rVSV-vectored vaccine in preventing Ebola virus disease: Final results from the Guinea ring vaccination, open-label, cluster-randomised trial (Ebola Ca Suffit!). Lancet 2017, 389, 505–518. [Google Scholar] [CrossRef]
  28. Wells, C.R.; Pandey, A.; Mbah, M.L.N.; Gaüzère, B.A.; Malvy, D.; Singer, B.H.; Galvani, A.P. The exacerbation of Ebola outbreaks by conflict in the Democratic Republic of the Congo. Proc. Natl. Acad. Sci. USA 2019, 116, 24366–24372. [Google Scholar] [CrossRef]
Figure 1. Study timeline.
Figure 1. Study timeline.
Vaccines 12 00828 g001
Figure 2. Disposition of delayed second dose immunogenicity subset participants.
Figure 2. Disposition of delayed second dose immunogenicity subset participants.
Vaccines 12 00828 g002
Figure 3. Ebola virus glycoprotein-specific binding antibody responses at pre-dose 2 and 21 days post-dose 2 in participants receiving a delayed dose 2 vaccine—distribution by age group. Antibody geometric mean concentration (GMC, ELISA Unit (EU)/mL) in adults (18 years or above), adolescents (12–17 years), and children (4–11 years). Boxes indicate median and interquartile range; EBOV (Ebola virus); LLOQ (Lower limit of quantification); ULOQ (Upper limit of quantification).
Figure 3. Ebola virus glycoprotein-specific binding antibody responses at pre-dose 2 and 21 days post-dose 2 in participants receiving a delayed dose 2 vaccine—distribution by age group. Antibody geometric mean concentration (GMC, ELISA Unit (EU)/mL) in adults (18 years or above), adolescents (12–17 years), and children (4–11 years). Boxes indicate median and interquartile range; EBOV (Ebola virus); LLOQ (Lower limit of quantification); ULOQ (Upper limit of quantification).
Vaccines 12 00828 g003
Figure 4. Immunogenicity of extended dose intervals for the Ad26.ZEBOV, MVA-BN-Filo vaccine regimen in this study and the EBL2002 trial. Closed circles depict immunogenicity results of healthy adult participants in the EBL2002 trial conducted in Burkina Faso, Côte D’Ivoire, Kenya, and Uganda who received dose 2 between 28 days to ≥280 days after dose 1. Open circles depict immunogenicity data collected in this DRC-EB-001 trial from immunogenicity subset adult participants in the DRC who received their dose 2 approximately 9.3 months after dose 1. A trendline (dashed line) is fitted by ordinary least square linear regression of EBL2002 data from healthy adults using STATA/SE 18.0.
Figure 4. Immunogenicity of extended dose intervals for the Ad26.ZEBOV, MVA-BN-Filo vaccine regimen in this study and the EBL2002 trial. Closed circles depict immunogenicity results of healthy adult participants in the EBL2002 trial conducted in Burkina Faso, Côte D’Ivoire, Kenya, and Uganda who received dose 2 between 28 days to ≥280 days after dose 1. Open circles depict immunogenicity data collected in this DRC-EB-001 trial from immunogenicity subset adult participants in the DRC who received their dose 2 approximately 9.3 months after dose 1. A trendline (dashed line) is fitted by ordinary least square linear regression of EBL2002 data from healthy adults using STATA/SE 18.0.
Vaccines 12 00828 g004
Table 1. Study participant demographic and baseline characteristics.
Table 1. Study participant demographic and baseline characteristics.
Immunogenicity Analysis PopulationAdults
(18+ Years)
Adolescents
(12–17 Years)
Children
(4–11 Years)
Participants with paired samples, n
(total = 133)
493252
Age (years)Mean (SD)31.0 (11.0)13.5 (1.3)6.8 (2.2)
Median (range)28.0
(18.0–62.0)
13.0
(12.0–17.0)
6.0
(4.0–11.0)
SexFemale, n (%)33 (67.3)15 (46.9)25 (48.1)
Male, n (%)16 (32.7)17 (53.1)27 (51.9)
Days between dose 1 and dose 2Mean (SD)265.4 (21.6)269.5 (16.1)258.0 (7.5)
Median (range)259.0
(244.0–338.0)
265.0
(249.0–327.0)
255.0
(248.0–283.0)
Months between dose 1 and dose 2Mean (SD)9.5 (0.8)9.6 (0.6)9.2 (0.3)
Median (range)9.3
(8.7–12.1)
9.5
(8.9–11.7)
9.1
(8.9–10.1)
SD, Standard deviation; range, minimum–maximum.
Table 2. Ebola virus glycoprotein-specific binding antibody concentrations.
Table 2. Ebola virus glycoprotein-specific binding antibody concentrations.
Immunogenicity Analysis PopulationAdults
(18+ Years)
Adolescents
(12–17 Years)
Children
(4–11 Years)
Participants with paired samples, n
(total = 133)
493252
Pre-dose 2
GMC (95% CI)
217
(157; 301)
378
(281; 510)
558
(471; 661)
21 days post-dose 2
GMC (95% CI)
22,194
(16,726; 29,449)
37,896
(29,985; 47,893)
34,652
(27,906; 43,028)
GMC fold change * between time points10210062
Data are number of paired samples (n), geometric mean concentration, and 95% confidence interval (GMC, 95% CI). * Fold change = GMC at 21 days post-dose 2/GMC at pre-dose 2.
Table 3. Comparison of Ebola virus glycoprotein-specific binding antibody concentrations between sub-Saharan African trials assessing the Ad26.ZEBOV, MVA-BN-Filo Ebola vaccine regimen.
Table 3. Comparison of Ebola virus glycoprotein-specific binding antibody concentrations between sub-Saharan African trials assessing the Ad26.ZEBOV, MVA-BN-Filo Ebola vaccine regimen.
CountriesDosing Interval Median Days (Range) *Number of Participants **Antibody GMC at Pre-dose 2Antibody GMC at 21 Days Post-Dose 2 GMC Fold Change after Dose 2 ††Trial Protocol [Reference]
Adults (18+ years)
DRC259
(244–338)
49217
(157; 301)
22194
(16,726; 29,449)
102DRC-EB-001
SL 56
(55–63)
54
(50–63)
42
(stage 1)
182
(stage 2)
269
(208; 347)
236
(206; 270)
4784
(3736; 6125)
3810
(3312; 4383)
18
16
EBL3001 [11]
BF/CI/KN/UG56
(54–58)
136361
(307; 423)
7518
(6468; 8740)
21EBL2002 [8]
Adolescents (12–17 years)
DRC264
(249–327)
32378
(281; 510)
37896
(29,985; 47,893)
100DRC-EB-001
SL 52
(50–58)
134314
(269; 366)
9929
(8172; 12,064)
32EBL3001 [17]
BF/CI/KN/UG 56
(53–57)
53562
(460; 686)
13532
(10,732; 17,061)
24EBL2002 [19]
Children (4–11 years)
DRC255
(248–283)
52558
(471; 661)
34652
(27,906; 43,028)
62DRC-EB-001
SL 55
(51–62)
124390
(334–456)
10212
(8419; 12,388)
26EBL3001 [17]
BF/CI/KN/UG 56
(54–59)
53658
(556–780)
17388
(12,973; 23,306)
26EBL2002 [19]
* The number of days between dose 1 and 2 expressed as median (minimum–maximum range). The data presented in this table include participants in previous randomised trials who received the Ebola vaccine regimen at the standard 56-day interval but exclude those who received placebo. The dosing interval medians and ranges reported here are therefore different from those published in previous papers whereby the placebo participants were also included. ** The number of participants with immunological data available at 21 days post-dose 2. GMC in [EU/mL] (95% CI). †† Fold change = GMC at 21 days post-dose 2/GMC at pre-dose 2. GMC, Geometric mean concentration; EU, ELISA Unit; BF, Burkina Faso; CI, Côte D’Ivoire; DRC, The Democratic Republic of the Congo; KN, Kenya; SL, Sierra Leone; UG, Uganda.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Choi, E.M.-L.; Kasonia, K.; Kavunga-Membo, H.; Mukadi-Bamuleka, D.; Soumah, A.; Mossoko, Z.; Edwards, T.; Tetsa-Tata, D.; Makarimi, R.; Toure, O.; et al. Immunogenicity of an Extended Dose Interval for the Ad26.ZEBOV, MVA-BN-Filo Ebola Vaccine Regimen in Adults and Children in the Democratic Republic of the Congo. Vaccines 2024, 12, 828. https://doi.org/10.3390/vaccines12080828

AMA Style

Choi EM-L, Kasonia K, Kavunga-Membo H, Mukadi-Bamuleka D, Soumah A, Mossoko Z, Edwards T, Tetsa-Tata D, Makarimi R, Toure O, et al. Immunogenicity of an Extended Dose Interval for the Ad26.ZEBOV, MVA-BN-Filo Ebola Vaccine Regimen in Adults and Children in the Democratic Republic of the Congo. Vaccines. 2024; 12(8):828. https://doi.org/10.3390/vaccines12080828

Chicago/Turabian Style

Choi, Edward Man-Lik, Kambale Kasonia, Hugo Kavunga-Membo, Daniel Mukadi-Bamuleka, Aboubacar Soumah, Zephyrin Mossoko, Tansy Edwards, Darius Tetsa-Tata, Rockyath Makarimi, Oumar Toure, and et al. 2024. "Immunogenicity of an Extended Dose Interval for the Ad26.ZEBOV, MVA-BN-Filo Ebola Vaccine Regimen in Adults and Children in the Democratic Republic of the Congo" Vaccines 12, no. 8: 828. https://doi.org/10.3390/vaccines12080828

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop